This invention relates to a taxoid of the formula (I): ##STR1## wherein R1 is a C3 -C5 alkyl or alkenyl or trifluoromethyl radical;

R2 is a C3 -C5 branched alkyl radical;

R3 and R4 are independently selected from hydrogen and hydroxyl protecting groups including functional groups which increase the water solubility of the taxoid antitumor agent;

R5 is a hydrogen, an acyl radical, or an alkoxylcarbonyl or carbamoyl radical; and

R6 is an acyl radical.

The compounds of formula I are useful as antitumor agents or their precursors. This invention also relates to a pharmaceutical composition having antineoplastic activity comprising the compound of formula (I) and a physiologically acceptable carrier and method of treatment using the compound of formula I.

Patent
   6100411
Priority
Oct 28 1994
Filed
Mar 04 1996
Issued
Aug 08 2000
Expiry
Oct 28 2014
Assg.orig
Entity
Small
18
5
all paid
1. A taxoid of the formula (I): ##STR10## wherein R1 is an unsubstituted or alkyl-substituted C3 alkyl or C3 alkenyl radical, wherein the C3 alkenyl radical is not a 1-propenyl radical;
R2 is a C3 -C5 branched alkyl radical;
R3 and R4 are hydrogen;
R5 is an acyl radical other than an acetyl radical, or an alkoxycarbonyl or carbamoyl radical; and
R6 is an acyl radical.
14. A taxoid of the formula (I): ##STR13## wherein R1 is a trifluoromethyl radical;
R2 is a C3 -C5 branched alkyl radical;
R3 and R4 are independently selected from hydrogen and hydroxyl protecting groups including functional groups which increase the water solubility of the taxoid antitumor agent;
R5 is hydrogen or an acyl, alkoxycarbonyl, or carbamoyl radical; and
R6 is an acyl radical.
2. A taxoid according to claim 1 wherein:
R1 is propyl, 2-methyl-1-propenyl, 1-methyl-1-propenyl, 2-methyl propyl, 1-methylpropyl, cyclopropyl, or cyclopropylmethyl radical;
R2 is selected from isopropyl, cyclopropyl, isobutyl, sec-butyl, 2-methylpropyl, 3-methylpropyl, tert-butyl, cyclobutyl, cyclopentyl, 1-ethylpropyl, or 1,1-dimethylpropyl radicals;
R5 is selected from C3 -C6 acyl, C1 -C6 alkoxylcarbonyl, C1 -C6 N-alkylcarbamoyl, or C1 -C6 N,N-dialkylcarbamoyl radicals;
R6 is selected from benzoyl, fluorobenzoyl, chlorobenzoyl, azidobenzoyl, cyclohexanecarbonyl, acryloyl, crotonoyl, 1-methylacryloyl, 2-methyl-2-butenoyl, or 3-methyl-3-butenoyl radicals; and
R3 and R4 are hydrogen.
3. A taxoid according to claim 1 wherein:
R1 is propyl, 2-methyl-1-propenyl, 1-methyl-1-propenyl, 2-methylpropyl, 1-methylpropyl, cyclopropyl, or cyclopropyl methyl radical;
R2 is a tert-butyl radical;
R5 is selected from triethoxycarbonyl, trifluoroacetyl, propanoyl, cyclopropanecarbonyl, acryloyl, crotonoyl, 3,3-dimethylacryloyl, N-methylcarbamoyl, N-ethylcarbamoyl, N-propylcarbamoyl, N-isopropylcarbamoyl, N-butylcarbamoyl, N-pentylcarbamoyl, N-hexylcarbamoyl, N, N-dimethylcarbamoyl, N, N-diethylcarbamoyl, N,N-dipropylcarbamoyl, N,N-dibutylcarbamoyl, pyrrolidine-N-carbonyl, piperidine-N-carbonyl, morpholine-N-carbonyl, methoxycarbonyl, ethoxylcarbonyl, propoxylcarbonyl, butoxycarbonyl, cyclopentanecarbonyl, or cyclohexanecarbonyl radicals; and
R6 is a benzoyl radical.
4. A taxoid according to claim 1 wherein:
R1 is propyl, 2-methyl-1-propenyl, 2-methylpropyl, cyclopropyl, or cyclopropylmethyl radical;
R2 is a tert-butyl radical;
R3 is hydrogen;
R4 is hydrogen;
R5 is selected from propanoyl, cyclopropanecarbonyl, acryloyl, crotonoyl, 3,3-dimethylacryloyl, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, pyrrolidine-N-carbonyl, piperidine-N-carbonyl, morpholine-N-carbonyl, methoxycarbonyl, ethoxylcarbonyl, propoxylcarbonyl, butoxycarbonyl, cyclopentanecarbonyl, or cyclohexanecarbonyl radicals; and
R6 is a benzoyl radical.
5. A taxoid according to claim 1 wherein:
R1 is a 2-methyl-1-propenyl, or 2-methylpropyl radical;
R2 is a tert-butyl radical;
R3 is hydrogen;
R4 is hydrogen;
R5 is selected from propanoyl, cyclopropanecarbonyl, crotonoyl, N,N-dimethylcarbamoyl, methoxycarbonyl, or acryloyl radicals; and
R1 is a benzoyl radical.
6. A pharmaceutical composition having antineoplastic activity comprising the compound of claim 1 and a physiologically acceptable carrier therefor.
7. A method for treating tumors which comprises administrating to a patient an effective antitumor amount of the compound of claim 1.
8. A method according to claim 7 wherein said treatment comprises treating tumors selected from the group consisting of leukemia, melanoma, breast, non-small cell lung, ovarian, and colon cancers.
9. A taxoid according to claim 1 wherein
R1 is a 2-methyl-1-propenyl or 2-methylpropyl radical,
R5 is an acyl radical other than an acetyl radical, alkoxycarbonyl, or N,N-dialkylcarbamoyl group, and
R2, R3, R4, and R6 are defined as in claim 1.
10. A method for preparing a taxoid according to claim 1 comprising coupling baccatins of formula (II) ##STR11## wherein G1 represents a hydroxyl protecting group, and R5 and R6 are defined in claim 1, with the β-lactams of formula (III) ##STR12## wherein G is a hydroxyl protecting group and R1 and R2 are defined in claim 1, in the presence of a base.
11. The taxoid according to claim 1, wherein:
R1 is a 2-methyl-1-propenyl or 2-methylpropyl radical; and
R2, R3, R4, R5, and R6 are defined as in claim 1.
12. The taxoid according to claim 1, wherein:
R1, R2, R3, R4, and R6 are defined as in claim 1; and
R5 is an acyl radical other than an acetyl radical, alkoxycarbonyl, or N,N-dialkylcarbamoyl group.
13. The taxoid according to claim 1, wherein the alkyl substituent of the C3 alkyl or C3 alkenyl radical is a methyl radical.
15. A method for treating tumors which comprises administering to a patient an effective amount of a taxoid of claim 14.
16. A pharmaceutical composition having antineoplastic activity comprising a taxoid of claim 14 and a physiologically acceptable carrier.

this application is a continuation-in-part of Ser. No. 08/330,956, filed Oct. 28, 1994, now abandoned.

The present invention relates to new taxoids possessing strong antitumor activities, the precursors of these antitumor taxoids, and pharmaceutical compositions thereof.

Taxol (paclitaxel), a complex diterpene, is currently considered the most exciting lead in cancer chemotherapy. Paclitaxel possesses high cytotoxicity and strong antitumor activity against different cancers which have not been effectively treated by existing antitumor drugs. For example, paclitaxel has been approved by FDA in late 1992 for the treatment of advanced ovarian cancer and for breast cancer in 1994. Paclitaxel is currently in phase II and III clinical trial for lung cancer and other cancers.

Although paclitaxel is an extremely important "lead" in cancer chemotherapy, it is common that better drugs can be derived from naturally occurring lead compounds. In fact, French researchers have discovered that a modification of the C-13 side chain of paclitaxel brought about a new anticancer agent which seems to have antitumor activity superior to paclitaxel with better bioavailability. This unnatural compound was named "Taxotere (docetaxel)", which has t-butoxycarbonyl instead of benzoyl on the amino group of (2R,3S)-phenylisoserine moiety at the C-13 position and a hydroxyl group instead of acetoxy group at C-10. Docetaxel is currently in phase II and III clinical trials in United States, Europe, and Japan, has shown excellent activity, especially against breast and lung cancers. ##STR2##

A recent report on clinical trials of paclitaxel and docetaxel has disclosed that paclitaxel causes, e.g., nerve damage, muscle pain or disturbances in heart rhythm, whereas docetaxel provokes, e.g., mouth sores and a plunge in white blood cells. Other less serious side effects also exist for these two drugs. Therefore, it is very important to develop new anti-cancer drugs different from these two drugs which have fewer undesirable side effects, better pharmacological properties, improved activity against drug-resistant tumors, and/or activity spectra against various tumor types.

It is an objective of the present invention to develop such new anti-tumor agents of paclitaxel class, i.e., taxoids, which have distinct structural differences from those of paclitaxel and docetaxel.

It is an object of the present invention to provide a series of new taxoids bearing a 1-propenyl, 2-methyl-1-propenyl, 2-methylpropyl, or trifluromethyl radical at the C-3' position instead of a phenyl group, and which possess strong antitumor activities with better therapeutic profile, in particular against drug-resistant tumors. One of the serious drawbacks of both paclitaxel and docetaxel is the fact that these two drugs possess only a weak activity against drug-resistant tumors, e.g., adriamycin-resistant breast cancer. The new taxoids of the present invention have shown not only stronger antitumor activities against human ovarian, non-small cell lung, colon, and breast cancers than those of the two drugs, but also exhibit more than one order of magnitude better activity against adriamycin-resistant human breast cancer cells than those of the two drugs. Multi-drug-resistance (MDR) is a serious issue in clinical oncology, and thus the new taxoid antitumor agents of this invention will serve as important drugs to overcome this problem.

A taxoid of the formula (I) ##STR3## in which R1 is a C3 -C5 alkyl or alkenyl or trifluoromethyl radical;

R2 is a C3 -C5 branched alkyl radical;

R3 and R4 are independently selected from hydrogen and hydroxyl protecting groups including functional groups which increase the water solubility of the taxoid antitumor agent;

R5 represents a hydrogen or hydroxyl-protecting an acyl or alkoxycarbonyl or carbamoyl group;

R6 represents an acyl radical, which are useful as antitumor agents or their precursors.

Preferably, R1 is selected from propyl, 2-methyl-1-propenyl, 1-methyl-1-propenyl, 2-methylpropyl, 1-methylpropyl, tert-butyl, cyclopropyl, cyclopropylmethyl, 1-methyl-1-butenyl, 2-methyl-1-butenyl, 3-methyl-1-butenyl, 1-methylbutyl, 2-methylbutyl, isobutyl, 2-methylethyl, 3-methylbutyl, 2-butenyl, or trifluoromethyl radicals;

R2 is selected from isopropyl, cyclopropyl, isobutyl, sec-butyl, 2-methylpropyl, 3-methylpropyl, tert-butyl, cyclobutyl, cyclopentyl, 1-ethylpropyl, or 1,1-dimethylpropyl radicals;

R5 is selected from hydrogen, C2 -C6 acyl, C1 -C6 alkoxylcarbonyl, C1 -C6 N-alkylcarbamoyl, or C1 -C6 N,N-dialkylcarbamoyl radicals; and

R6 is selected from benzoyl, fluorobenzoyl, chlorobenzoyl, azidobenzoyl, cyclohexanecarbonyl, acryloyl, crotonoyl, 1-methylacryloyl, 2-methyl-2-butenoyl, or 3-methyl-3-butenoyl radical.

More preferably, R5 is selected from acetyl, propanoyl, cyclopropanecarbonyl, acryloyl, crotonoyl, 3,3-dimethylacryloyl, N-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, N,N-diethylcarbamoyl, pyrrolidine-N-carbonyl, piperidine-N-carbonyl, morpholine-N-carbonyl, methoxycarbonyl, ethoxylcarbonyl, propoxylcarbonyl, butoxycarbonyl, cyclopentanecarbonyl, or cyclohexanecarbonyl radicals.

These new taxoids (I) are synthesized by the processes which comprise the coupling reactions of the baccatin of the formula (II) ##STR4## wherein G1 represents a hydroxyl protecting group, and R5 and R6 have been defined above, with the β-lactams of the formula (III) ##STR5## wherein G is a hydroxyl protecting group such as ethoxyethyl (EE), triethylsilyl (TES), (tert-butyl)dimethylsilyl (TBS), and triisopropylsilyl (TIPS), and R1 and R2 have been defined above, in the presence of a base.

New taxoids of the formula (I) hereinabove are useful as antitumor agents or their precursors. These taxoids possess strong antitumor activities against human breast, non-small cell lung, ovarian, and colon cancers including drug-resistant cancer cells, as well as leukemia and melanoma.

The new taxoids of the formula (I) are synthesized by modifying the baccatins of the formula (II) ##STR6## wherein G1, R5, and R6 have been defined above.

The baccatins (II) are coupled with the β-lactams of the formula (III) ##STR7## wherein G, R1, and R2 have been defined hereinabove, to yield the new taxoids (I)

The β-lactams (III) are readily prepared via the β-lactams (IV) which are easily obtained through the chiral enolate-imine cyclocondensation method that has been developed in the present inventor's laboratory as shown in Scheme 1 (Ojima et al., Bioorg. Med. Chem. Lett., 1993, 3, 2479, Ojima et al., Tetrahedron Lett., 1993, 34, 4149, Ojima et al., Tetrahedron Lett. 1992, 33, 5739, Ojima et al., Tetrahedron, 1992, 48, 6985, Ojima, I. et al., J. Org. Chem., 1991, 56, 1681, the disclosures of which are incorporated herein by reference). In this preparation, the β-lactams (IV) with extremely high enantiomeric purities are obtained in high yields. In Scheme 1, R* is a chiral auxiliary moiety which is (-)-trans-2-phenyl-1-cyclohexyl or (-)-10-dicyclohexylsulfamoyl-D-isobornyl, TMS is a trimethylsilyl radical, and the base is lithium diisopropylamide or lithium hexamethyldisilazide and G and R1 have been defined hereinabove. ##STR8##

The β-lactams (IV) can be converted to the corresponding N-alkoxycarbonyl-β-lactams (III) in excellent yields by reacting with alkyl chloroformates in the presence of a base (Scheme 2). This transformation is known to those skilled in the art.

The β-lactams (III) are readily used for the coupling with the baccatins (II) in the presence of a base, followed by deprotection to give the new taxoids (I) in high yields (Scheme 3). ##STR9##

In Schemes 1-3, R1 through R6 have been defined above, M is an alkali metal, and the hydroxyl protecting group G1 is independently selected from methoxylmethyl (MOM), methoxyethyl (MEM), 1-ethoxyethyl (EE), benzyloxymethyl, (β-trimethylsilylethoxyl)-methyl, tetrahydropyranyl, 2,2,2-trichloroethoxylcarbonyl (Troc), benzyloxycarbonyl (CBZ), tert-butoxycarbonyl (t-BOC), 9-fluorenylmethoxycarbonyl (Fmoc), 2,2,2-trichloroethoxymethyl trimethylsilyl, triethylsilyl, tripropylsilyl, dimethylethylsilyl, dimethyl(t-butyl)silyl, diethylmethylsilyl, dimethylphenylsilyl, diphenylmethylsilyl, acetyl, chloroacetyl, dichloroacetyl, trichloroacetyl and trifluoroacetyl.

The coupling reaction of the baccatin (II) and the β-lactams (VI) is carried out via an alkali metal alkoxide of the baccatin (II) at the C-13 hydroxyl group. The alkoxide can readily be generated by reacting the baccatin with an alkali metal base such as sodium hexamethyldisilazide, potassium hexamethyldisilazide, lithium hexamethyldisilazide, sodium diisopropylamide, potassium diisopropylamide, lithium diisopropylamide, sodium hydride, in a dry nonprotic organic solvent such as tetrahydrofuran (THF), dioxane, ether, dimethoxyethane (DME), diglyme, dimethylformamide (DMF), mixtures of these solvents with hexane, toluene, and xylene, in a preferred temperature range from about -100°C to about 50° C., more preferably at about -78°C to about 25°C This reaction is preferably carried out under inert atmosphere such as nitrogen and argon. The amount of the base used for the reaction is preferably approximately equivalent to the amount of the baccatin when soluble bases such as sodium hexamethyldisilazide, potassium hexamethyldisilazide, lithium hexamethyldisilazide, sodium diisopropylamide, potassium diisopropylamide, lithium diisopropylamide are used. The use of a slight excess of the base does not adversely affect the reaction. When heterogeneous bases such as sodium hydride and potassium hydride are used, 5-10 equivalents of the base (to the amount of the baccatin) are preferably employed.

The coupling reaction of the metal alkoxide of the baccatin thus generated with the β-lactam is typically carried out by adding the solution of the β-lactam in a dry organic solvent exemplified above in a preferred temperature range from about -100°C to 50°C, more preferably at about -35°C to 25°C The mixture of reactants is stirred for 15 minutes to 24 hours and the progress and the completion of the reaction is monitored by thin layer chromatography (TLC), for example. When the limiting reactant is completely consumed, the reaction is quenched by addition of a cold brine solution. The crude reaction mixture is worked up using the standard isolation procedures which are generally known to those skilled in the art to give the corresponding taxoid. The proportion of the β-lactam and the baccatin is in a range from 2:1 to 1:2, more preferably approximately 1:1 for purposes of economy and efficiency, but the ratio is not critical for the reaction.

The hydroxyl protecting groups can then be removed by using the standard procedures which are generally known to those skilled in the art to give the desired taxoid derivatives. For example, EE and TES groups can be removed with 0.5 N HCl at room temperature for 36 h, TIPS and TBS groups can be removed by treating with fluoride ion or HF in a non-protic organic solvent, and Troc group can be removed with zinc and acetic acid in methanol at 60°C for 1 hour without disturbing the other functional groups and the skeleton of the taxoid.

It has been shown that the introduction of 2-methyl-1-propenyl group to the C-3' position of paclitaxel appears to increase the cytotoxicity, especially against drug-resistant cancer cells: Holton and Nadizadeh disclosed in U.S. Pat. No. 5,284,864 (1994) that 3'-desphenyl-3'-isobutenylpaclitaxel (RAH-1) exhibited 4 times better activity than paclitaxel and 7 times better activity than docetaxel against human colon carcinoma cells HCT-116, and also about 20 times better activity than paclitaxel and 9 times better activity than docetaxel against multi-drug resistant phenotype human colon carcinoma cells HCT-116/VM.

We have found that the structural requirements for taxoid antitumor agents to express strong potency are rather strict and unpredictable. For example, 3'-desphenyl-3'-(2-phenylethenyl)docetaxel, bearing 2-phenylethenyl group instead of the isobutenyl group of RAH-1, has dramatically decreased cytotoxicity (>20 times) and 3'-desphenyl-3'-neopentyldocetaxel, bearing neopentyl group which has just one more methyl than isobutenyl group, is virtually not cytotoxic against A121 human ovarian, A549 human non-small cell lung, HT-29 human colon and MCF7 human breast cancer cells. While looking at the structure-activity relationships (SAR) of new taxoids that have different substituents at the C-3' and C-10, we discovered that there are optimum combinations of these two substituents which achieve extraordinarily high activity against drug-resistant cancer cells.

After searching for the best substituent for the C-3' and the C-10 positions by employing many alkyl groups and alkenyl groups by trial and error, we have identified 1-propenyl, 2-methyl-1-propenyl, 2-methylpropyl, and trifluoromethyl groups to be the optimum substituents for the C-3' position, and acyl groups, alkoxycarbonyl groups, and N,N-dialkylcarbamoyl groups to be the optimum substituents for the C-10 position.

For example, 3'-desphenyl-3'-(1-propenyl)-10-acetyldocetaxel (Taxoid Ia) showed a substantially better activity spectrum than that of paclitaxel and docetaxel against human ovaian, human non-small cell lung, human colon, and human breast cancer cells mentioned above (see TABLE 1 in EXAMPLE 32). Moreover, this agent possesses 21 times better activity than paclitaxel and 17 times better activity than docetaxel against the drug-resistant human breast cells MCF7-R, which are mammary carcinoma cells 180 fold resistant to a widely used anticancer drug, adriamycin. In the same assay, Holton's compound RAH-1 showed only marginal activity that was one order of magnitude weaker than that of Taxoid Ia (see TABLE 1 in EXAMPLE 32).

3'-Desphenyl-3'-(2-methyl-1-propenyl)-10-cyclopropanecarbonyldocetaxel (Taxoid IX) showed one order of magnitude better activity than that of paclitaxel and docetaxel against human human breast cancer cells mentioned above (see TABLE 2 in Example 32), and possesses two order of magnitude (142 times) better activity against the drug-resistant human breast cells mentioned above. These extraordinarily high activities are totally unpredictable from the exsisting SAR studies of paclitaxel and docetaxel, and thus demonstrate the exceptional importance of our discovery.

The taxoids of the formula (I) of this invention are useful for inhibiting tumor growth or regression of tumors in animals including humans and are preferably administered in the form of a pharmaceutical composition including effective amounts of the antitumor agent of this invention in combination with a pharmaceutically acceptable vehicle or diluent.

The pharmaceutical compositions of the antitumor agents of the present invention may be made in any form suitable for desired use, e.g., oral, parenteral or topical administration. Examples of parenteral administration are intramuscular, intraveneous, intraperitoneal, rectal, and subcutaneous administration. The vehicle or diluent ingredients should not reduce the therapeutic effects of the antitumor agents of this invention.

Suitable dosage forms for oral use include tablets, dispersible powders, granules, capsules, suspension, syrups, and elixirs. Examples of inert diluents and vehicles for tablets include calcium carbonate, sodium carbonate, lactose and talc. Examples of inert diluents and vehicles for capsules include calcium carbonate, calcium phosphate, and kaolin. Dosage forms appropriate for parenteral administration include solutions, suspensions, dispersions, and emulsions.

The water solubility of the antitumor agents of the formula (I) may be improved by modifying the C-2' and /or C-7 substituents to incorporate suitable functional groups, such as R3 and R4. In order to increase the water solubility, R3 and R4 can be independently selected from hydrogen and --CO--X--Y, wherein X is selected from --(CH2)n -- (n=1-3), --CH═CH--, cyclohexanediyl, and benzenediyl and Y is selected from --COOH and its pharmaceutically acceptable salts, --SO3 H and its pharmaceutically acceptable salts, --NR7 R8 and its pharmaceutically acceptable salts, the pharmaceutically acceptable ammonium salt --NR7 R8 R9, --CONR8 R9, or --COOR9, in which

--NR7 R8 includes cyclic amine radicals selected from pyrrolidinyl, piperidinyl, morphorino, piperazinyl, and N-methylpiperazinyl;

R7 and R8 are independently selected from hydrogen, allyl, C1 -C6 alkyl, and --(CH2)n --Z (n=1-3);

R9 is selected from C1 -C6 alkyl, allyl, and --(CH2)n --Z (n=1-3), and

Z is selected from --COOH and its pharmaceutically acceptable salts, --SO3 H and its pharmaceutically acceptable salts, --NR7 R8 and its pharmaceutically acceptable salts, and pharmaceutically acceptable ammonium salt --NR7 R8 R10, in which R10 is selected from hydrogen, allyl, and C1 -C6 alkyl.

The preparation of the water-soluble analogs of paclitaxel bearing the functionalized acyl groups described above has been disclosed in Kingston et al., U.S. Pat. No. 5,059,699 (1991); Stella et al., U.S. Pat. No. 4,960,790 (1990), the disclosures of which are incorporated herein by reference, and thus it is not difficult for the people in the art to carry out such modifications.

The following non-limiting examples are illustrative of the present invention. It should be noted that various changes could be made in the examples and processes therein without departing from the scope of the present invention. For this reason, it is intended that the illustrative embodiments of the present application should be interpreted as being illustrative and not limiting in any sense.

(-)-(1R,2S)-2-phenyl-1-cyclohexyl triisopropylsilyloxyacetate:

A solution of (-)-(1R,2S)-2-phenyl-1-cyclohexyl hydroxyacetate (851 mg, 3.63 mmol) was prepared through esterification of benzyloxyacetyl chloride with (-)-(1R,2S)-2-phenyl-1-cyclohexanol followed by hydrogenolysis. Then, triisopropylsilyl chloride (840 mg, 4.36 mmol) and imidazole (618 mg, 9.08 mmol) in dimethylformamide (DMF) (1.7 mL) was added and stirred at room temperature for 12-20 hours. The mixture was poured into pentane (25 mL), and washed with water and brine. The combined organic layers were dried over anhydrous MgSO4 and concentrated in vacuo. The crude product was subjected to a purification on a short silica gel column using hexane/chloroform (3/1) as the eluant to give pure (-)-(1R,2S)-2-phenyl-1-cyclohexyl triisopropylsilyloxyacetate (1.35 g, 95% yield) as a colorless oil: [α]D20 -17.1° (c 3.15, CHCl3), IR (neat) 1759, 1730 (n CO) cm-1 ; 1 H NMR (CDCl3) δ0.93-0.99 (m, 21H), 1.30-1.62 (m, 4H), 1.72-2.0 (m, 3H), 2.10-2.19 (m, 1H), 266 (dt, J=11.5, 4.0 Hz, 1H), 3.90 (d, J=16.6 Hz, 1H), 4.07 (d, J=16.6 Hz, 1H), 5.07 (dt, J=10.6, 4.0 Hz, 1H), 7.16-7.30 (m, 5H). Anal. Calcd for C23 H38 O3 Si: C, 70.72; H, 9.81 Found: C, 70.79; H, 9.85.

N-(4-Methoxyphenyl)-2-alkenylaldimine:

To a solution of 0.360 g. (2.9 mmol) of p-anisidine (recrystallized twice from ethanol) in 12 mL of CH2 Cl2 over anhydrous Na2 SO4 was added 0.24 g (3.5 mmol) of 2-butenal (crotonaldehyde) (distilled immediately prior to use) under nitrogen. After 4 hours, Na2 SO4 was filtered off and the solvent removed under vacuum to give N-(4-methoxyphenyl)-2-butenaldimine in quantitative yield, which was used for the synthesis of β-lactam without further purification.

In the same manner, N-(4-methoxyphenyl)-3-methyl-22-butenaldimine was obtained in quantitative yield.

(3R,4S)-1-(4-Methoxyphenyl)-3-triisopropylsilyloxy-4-(1-alkenyl)azetidin-2- one (V):

To a solution of 0.27 mL (1.9 mmol) of diisopropylamine in 10 mL of THF was added 0.76 mL (1.9 mmol) of 2.5M n-butyllithium in hexanes at -10° C. After stirring for 45 minutes, the solution was cooled to -85° C. A solution of (-)-(1R,2S)-2-phenyl-1-cyclohexyl triisopropylsilyloxy-acetate (0.575 g 1.47 mmol) in 10 mL of THF was added via cannula over a period of 1.5 hours. After stirring for an additional hour, a solution of N-(4-methoxyphenyl)-2-butenaldimine (336 mg, 2.2 mmol) in 10 mL of THF was added via cannula over a period of approximately 1 hour. The mixture was stirred for 2 hours and allowed to warm up to room temperature overnight while stirring. The reaction was then quenched with saturated NH4 Cl. The aqueous layer was extracted with ethyl acetate (EtOAc) and the combined organic layers were washed with saturated NH4 Cl solution, and brine, and then dried over MgSO4. After the removal of solvent under vacuum, the crude product was obtained, which was purified by flash chromatography on silica gel (hexane:EtOAc=10:1 to 6:1) to afford pure PMP-β-lactam Va (399 mg, 70% yield) as a rust-colored oil. The enantiomeric purity of the PMP-β-lactam Va was determined to 97% ee on the basis of chiral HPLC analysis: [α]D =+33.1° (c 0.27, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ1.04-1.16 (m, 21H), 1.76 (dd, J=6.5, 1.3 Hz, 3H), 3.74 (s, 3H), 4.51 (dd, J=8.6, 5.0 Hz, 1H), 5.04 (d, J=5.0 Hz, 1H), 5.59 (ddd, J=15.4, 8.6, 1.3 Hz, 1H), 5.92 (dq, J=15.4, 6.5 Hz, 1H), 6.83 (d, J=9.0, 2H), 7.36 (d, J=9.0 Hz, 2H); 13 C NMR (63 MHz, CDCl3) d 11.89, 17.63, 17.68, 55.38, 61.89, 77.57, 114.18, 118.48, 126.65, 127.48, 128.34, 128.55, 132.59, 156.03. 165.43.

In the same manner, PMP-β-lactam Vb (R1 =2-methyl-1-propenyl) was obtained in 73% yield (93% ee): [α]D =+65.7° (c 1.00, CHCl3); 1 H NMR (CDCl3, 300 MHz) δ1.08-1.12 (m, 21 H), 1.81 (s, 3 H), 1.86 (s, 3 H), 3.78 (s, 3 H), 4.79-4.84 (dd, J=5.1, 9.9 Hz, 1 H), 5.05-5.07 (d, J=5.1 Hz, 1 H), 5.33-5.36 (bd, J=9.9 Hz, 1 H); 13 C NMR (CDCl3, 63 MHz) d 11.92, 17.61, 18.33, 26.11, 55.44, 57.56, 76.51, 77.015, 77.52, 114.25, 118.34, 120.15, 128.73, 131.52, 139.14, 156.00, 165.61.

(3R,4S)-3-Triisopropylsilyloxy-4-(1-alkenyl)azetidin-2-one (IV):

To a solution of 260 mg. (0.67 mmol) of N-PMP-β-lactam Va in 20 ml. of acetonitrile at -10°C, was added dropwise a solution of 1.13 g (2.07 mmol) of cerium ammonium nitrate (CAN) in 25 mL of water. The mixture was allowed to stir for 1 hour and then diluted with 50 mL of water. The aqueous layer was extracted with ethyl acetate (2×35 mL) and the combined organic layers were washed with water, 5% NaHSO3, 5% Na2 CO3, and brine. After drying over MgSO4 and concentrating under vacuum, the organic layers afforded the crude product, which was purified on a silica gel column using hexane-ethyl acetate as the eluant (hexane:EtOAc=3:1) to give the pure β-lactam IVa (R1 =1-propenyl) (124 mg, 65% yield) as a pale yellow viscous oil: 1 H NMR (CDCl3, 250 MHz) δ1.04-1.16 (m, 21H), 1.70 (dd, J=6.5, 1.2 Hz, 3H), 4.13 (dd, J=8.7, 4.9, 1H), 4.94 (d, J=4.9 Hz, 1H), 5.51 (ddd, J=14.1, 8.7, 1.2 Hz, 1H), 5.67 (m, 1H), 6.68 (br s, 1H); 13 C NMR (63 MHz, CDCl3) d 11.80, 17.57, 17.62, 58.14, 79.18, 127.97, 130.64, 170.36.

In the same manner, β-lactam IVb (R1 =2-methyl-1-propenyl) was obtained in 94% yield: 1 H NMR (CDCl3, 300 MHz) δ1.02-1.10 (m, 21 H), 1.65 (s, 3 H), 1.72 (s, 3 H), 4.36-4.40 (dd, J=4.5, 9.6 Hz, 1 H), 4.91-4.93 (dd, J=2.1, 4.5 Hz, 1H), 5.23-5.26 (bd, J=9.6 Hz, 1 H), 6.28 (bs, 1 H, NH).

(3R,4S)-1-tert-Butoxycarbonyl-3-triisopropylsilyloxy-4-(1-alkenyl)azetidin- 2-one (III):

To a solution of 100 mg (0.35 mmol) of the β-lactam IVa, 0.24 mL (1.75 mmol) of triethylamine, and a catalytic amount of dimethylaminopyridine (DMAP) in 11 mL of CH2 Cl2, was added dropwise at room temperature, 85 mg. (0.38 mmol) of di(tert-butyl) dicarbonate in 2 mL of CH2 Cl2. The mixture was stirred for 1 hour and quenched with saturated NH4 Cl solution. The mixture was diluted with 60 mL of ethyl acetate and the organic layer was washed with brine, dried over MgSO4, and concentrated. The crude product was purified by flash chromatography on silica gel (hexane:EtOAc=4:1) to yield pure N-t BOC-β-lactam IIIa (R1 =1-propenyl) as colorless oil (105 mg, 87% yield): 1 H NMR (CDCl3, 250 MHz) δ1.02-1.08 (m, 21H), 1.48 (s, 9H), 1.74 (dd, J=6.4, 1.3 Hz, 3H), 4.44 (dd, J=8.6, 5.8 Hz, 1H), 4.94 (d, J=5.8 Hz, 1H), 5.54 (ddd, J=15.4, 8.6, 1.3 Hz), 5.83 (dq, J=15.4, 6.4 Hz, 1H); 13 C NMR (63 MHz, CDCl3) δ11.76, 17.52, 17.95, 27.97, 61.04, 83.06, 124.80, 132.72, 148.0, 166.07. Anal. Calcd for C20 H37 NO4 Si: C, 62.62, H, 9.72, N, 3.65. Found: C, 62.62; H, 9.63; N, 3.61.

In the same manner, N-t BOC-β-lactam IIIb (R1 =2-methyl-1-propenyl) was obtained as a colorless oil in 82% yield: 1 H NMR (CDCl3, 300 MHz) δ0.97-1.06 (m, 21 H), 1.48 (s, 9 H), 1.75 (s, 3 H), 1.78 (s, 3 H), 4.72-4.77 (dd, J=5.7, 9.9 Hz, 1 H), 4.94-4.96 (dd, J=5.7 Hz, 1 H), 5.25-5.28 (bd, J=9.9 Hz , 1 H).

7-Triethylsilyl-10-O-substituted 10-deacetylbaccatin III (IIb-g).

To a solution of 1.0 g (1.84 mmol) of 10-deacetylbaccatin III and 375 mg (5.52 mmol) of imidazole in 10 mL DMF was added dropwise 0.9 mL (5.52 mmol) of chlorotriethylsilane (TESCl). The reaction mixture was stirred for 5 hours at room temperature and quenched with water, then diluted with EtOAc. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with water, dried over MgSO4, and concentrated. The crude product was purified by flash chromatography on silica gel (hexane/EtOAc, 1:1) to give 774 mg (64%) of 7-triethylsilyl-10-deacetylbaccatin III (7-TES-DAB) as a white solid: 1 H NMR (CDCl3, 250 MHz) δ0.50 (m, 6 H), 0.97 (m, 9 H), 1.21 (s, 3 H), 1.58 (s, 3 H), 1.73 (s, 3 H), 1.85 (dt, 1 H), 1.99 (s, 3 H), 2.23 (s, 3 H), 2.24 (s, 2 H), 2.47 (ddd, 1 H), 3.94 (d, J=7.2 Hz, 1 H), 4.14 (AB, JAB =8.4 Hz, 1 H), 4.32 (AB, JAB =8.1 Hz, 1 H), 4.41 (d, J=6.3 Hz, 1 H), 4.84 (t, 1 H), 4.94 (d, J=8.4 Hz, 1 H), 5.14 (s, 1 H), 5.19 (s, 1 H), 5.58 (d, J=7.2 Hz, 1 H), 7.40 (t, 2 H), 7.54 (t, 1 H), 8.10 (d, 2 H).

To 77 mg (0.117 mmol) of 7-TES-DAB in 5 mL THF was added 0.12 mL of LiHMDS (1M in THF). The reaction mixture was stirred at -40°C for 5 minutes, then 0.010 mL (0.117 mmol) of propanoyl chloride (previously distilled) was added. The solution was allowed to warm up at 0°C over a 30 min period. Then the solvent was removed in vacuo and the crude product was purified by flash chromatography on silica gel (hexane then hexane/EtOAc, 4:1, then 2:1 and 1:1) to afford 60 mg (72%) of 7-triethylsilyl-10-propanoyl-10-deacetylbaccatin III (IIb) as a white solid: [α]D21 -68.57° (c 1.75, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ0.52 (q, 6 H), 0.87 (t, 9 H), 1.02 (s, 3 H), 1.16-1.22 (m 6 H), 1.55 (s, 9 H), 1.67 (s, 3 H), 1.86 (m, 1 H), 2.19 (s, 3 H), 2.25 (s, 2 H), 2.27 (s, 3 H), 2.42 (m, 3 H), 3.86 (d, J=6.9 Hz, 1 H), 4.12 (AB, JAB =8.0 Hz, 1 H), 4.27 (AB, JAB =8.0 Hz, 1 H), 4.49 (dd, 1 H), 4.83 (t, 1 H), 4.93 (d, J=9.2 Hz, 1 H), 5.61 (d, J=6.9 Hz, 1 H), 6.46 (s, 1 H), 7.43 (t, 2 H), 7.56 (t, 1 H), 8.08 (d, 2 H); 13 C NMR (CDCl3, 63 MHz) δ5.2, 6,7, 9.2, 9.9, 14.9, 20.1, 22.6, 26.7, 27.6, 37.2, 38.3, 42.7, 58.6, 67.8, 72.3, 74.7, 75.5, 76.5. 77.0, 77.5, 78.7, 80.8, 84.2, 128.5, 129.4, 130.0, 132.6, 133.5, 143.9, 167.8, 170.7, 174.5, 202.3. IR (neat, cm-1) 2953, 2913, 1789, 1738, 1715, 1681, 1454, 1434, 1392, 1362, 1315, 1175, 1108. HRMS (FAB, DCM/NBA) m/z: Calcd. for C38,H54 O11 SiH+, 715.3513. Found, 715.3552.

In the same manner, the following 7-triethylsilyl-1-O-substituted-10-deacetylbaccatin IIIs were prepared.

7-Triethylsilyl-10-cyclopropanecarbonyl-10-deacetylbaccatin III (IIc).

White solid; [α]D21 -61.42° (c 7.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ0.46 (m, 6 H), 0.82 (m, 9 H), 0.97 (s, 3 H), 1.12 (s, 3 H), 1.18 (m, 2 H), 1.60 (s, 3 H), 1.68 (m, 2 H), 1.79 (m, 1 H), 2.12 (s, 3 H), 2.16 (s, 2 H), 2.20 (s, 3 H), 2.40 (m, 1 H), 2.50 (d, 1 H), 3.79 (d, J=6.9 Hz, 1 H), 4.03 (AB, JAB =8.1 Hz, 1 H), 4.24 (AB, JAB =8.1 Hz, 1 H), 4.38 (DD. J=6.6 Hz, 10.1 Hz, 1 H), 4.75 (t, 1 H), 4.87 (d, J=9.0 Hz, 1 H), 5.55 (d, J=6.6 Hz, 1 H), 6.39 (s, 1 H), 7.37 (t, 2 H), 7.51 (t, 1 H), 8.02 (d, 2 H); IR (neat, cm-1) 2958, 2356, 1771, 1732, 1716, 1699, 1652, 1558, 1456, 1393, 1268, 1169, 1107, 1070, 1026, 738. Anal. Calcd. for C39 H54 O11 Si: C, 64.44; H, 7.49. Found: C, 64.52, H, 7.49.

7-Triethylsilyl-10-crotonoyl-10-deacetylbaccatin III (IId).

White solid; [α]D22 -68.57° (c 7.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ0.51 (q, 6 H), 0.86 (t, 9 H), 1.00 (s, 3 H), 1.20 (s, 3 H), 1.66 (s, 3 H), 1.80 (m, 1 H), 1.88 (d, 3 H), 2.19 (s, 2 H), 2.20 (s, 3 H), 2.26 (s, 3 H), 2.51 (m, 3 H), 3.87 (d, J=6.8 Hz, 1 H), 4.80 (AB, JAB =8.2 Hz, 1 H), 4.26 (AB, JAB =8.2 Hz, 1 H), 4.45 (dd, J=6.7 Hz, 9.9 Hz, 1 H), 4.80 (T, 1 H), 4.92 (d, J=9.7 Hz, 1 H), 5.61 (d, J=6.8 Hz, 1 H), 5.92 (d, J=15 Hz, 1 H), 6.48 (s, 1 H), 7.02 (m, 1 H), 7.42 (t, 2 H), 7.55 (t, 1 H), 8.07 (d, 2 H); 13 C (CDCl3, 63 MHz) δ5.2, 6.7, 9.9, 14.9, 18.1, 20.1, 22.6, 26.7, 37.2, 38.2, 42.7, 47.3, 58.6, 67.9, 72.3, 74.7, 75.5, 76.5, 77.0, 77.5, 78.7, 80.8, 84.2, 122.3, 128.5, 129.4, 130.0, 132.7, 133.6, 143.9, 145.6, 164.7, 167.1, 170.7, 202.3; IR (neat, cm-1) 2953, 2356, 1716, 1558, 1455, 1267, 1173, 1106, 1001, 822.

7-Triethylsilyl-10-N,N-dimethylcarbamoyl-10-deacetylbaccatin III (IIe).

White solid; [α]D21 -30° (c 2.00, CHCl3); 1 H NMR (CDCl3, 300 MHz) δ0.57 (m, 6 H), 0.91 (m, 9 H), 1.21 (s, 3 H), 1.27 (s, 3 H), 1.69 (s, 3 H), 1.84 (dt, 1 H), 2.21 (s, 2 H), 2.26 (s, 3 H), 2.29 (s, 2 H), 2.49 (m, 1 H), 2.95 (s, 3 H), 3.09 (s, 3 H), 3.91 (d, J=6.9 Hz, 1 H), 4.12 (AB, JAB =8.4 Hz, 1 H), 4.30 (AB, JAB =8.4 Hz, 1 H), 4.48 (dd, J=6.7 Hz, 10.2 Hz, 1 H), 4.84 (t, 1 H), 4.97 (d, J=9.0 Hz, 1 H), 5.64 (d, J=6.9 Hz, 1 H), 6.40 (s, 1 H), 7.46 (t, 2 H), 7.59 (t, 1 H), 8.11 (d, 2 H). HRMS (FAB, DCM/NBA/NaCl) m/z: Calcd. for C38 H55 O11 NSiNa+, 752.3442. Found, 752.3483.

7-Triethylsilyl-10-methoxycarbonyl-10-deacetylbaccatin III (IIf).

White solid; [α]D22 -72.50° (c 4.00, CHCl3); 1 H NMR (CDCl3, 300 MHz) δ0.54 (M, 6 H), 0.89 (m, 9 H), 1.03 (s, 3 H), 1.15 (s, 3 H), 1.67 (s, 3 H), 1.82 (dt, 1 H), 2.18 (s, 3 H), 2.25 (s, 2 H), 2.27 (s, 3 H), 2.47 (ddd, 1 H), 3.82 (s, 3 H), 3.84 (d, 1 H), 4.11 (AB, JAB =8.1 Hz, 1 H), 4.27 (AB, JAB =8.1 Hz, 1 H), 4.44 (dd, J=6.6 Hz, 10.2 Hz, 1 H), 4.83 (t, 1 H), 4.93 (d, J=9.0 Hz, 1 H), 5.59 (d, J=6.9 Hz, 1 H), 6.27 (s, 1 H), 7.43 (t, 2 H), 7.56 (t, 1 H), 8.07 (d, 2 H). IR (neat, cm-1) 3524, 2957, 1715, 1442, 1371, 1266, 1108, 1025, 912, 820, 732.

7-Triethylsilyl-10-acryloyl-10-deacetylbaccatin III (IIg).

White solid; [α]D22 -77.5° (c 4.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ0.51 (q, 6 H), 0.87 (t, 9 H), 1.01 (s, 3 H), 1.21 (s, 3 H), 1.68 (s, 3 H), 1.81 (m, 1 H), 2.15 (d, 2 H), 2.22 (s, 3 H), 2.27 (s, 3 H), 2.46 (m, 1 H), 3.87 (d, J=6.9 Hz, 1 H), 4.12 (AB, JAB =8.3 Hz, 1 H), 4.28 (AB, JAB =8.3 Hz, 1 H), 4.47 (dd, J=6.7 Hz, 10.2 Hz, 1 H), 4.81 (t, 1 H), 4.94 (d, J=8.7 Hz, 1 H), 5.62 (d, J=6.9 Hz, 1 H), 5.88 (d, J=10.7 Hz, 1 H), 6.18 (m, 1 H), 6.47 (m, 1 H), 6.51 (s, 1 H), 7.02 (m, 1 H), 7.43 (t, 2 H), 7.56 (t, 1 H), 8.08 (d, 2 H); 13 C NMR (CDCl3, 63 MHz) δ5.2, 6,7, 9.9, 14.9, 20.1, 22.6, 26.7, 37.2, 38.2, 42.7, 47.3, 58.6, 67.9, 72.3, 74.7, 75.8, 76.5, 77.0, 77.5, 78.7, 80.8, 84.2, 128.1, 128.5, 129.3, 130.0, 131.5, 132.5, 133.6, 144.2, 164.5, 167.0, 170.7, 202.0; IR (neat, cm-1) 2950, 2250, 1734, 1717, 1653, 1635, 1506, 1457, 1362, 1269.

7-Triethylsilyl-10-O-substituted 2'-triisopropylsilyl-3'-(1-propenyl)docetaxel (I-P):

To a solution of 68 mg (0.097 mmol) of 7-triethylsilylbaccatin III (IIa) and 58 mg (0.15 mmol) of the N-t BOC-β-lactam (VIa) in 4 mL of THF at -30°C was added 0.12 mL (0.12 mmol) of LiHMDS. The mixture was allowed to warm to -10°C and stirred for 1 hour and was then quenched with NH4 Cl. The aqueous layer was extracted with 75 mL of EtOAc and the combined organics were washed with NH4 Cl and brine. The organics were then dried over MgSO4 and concentrated under vacuum. Upon purification by flash column chromatography on silica gel (hexane:EtOAc=4:1), 83 mg (79% yield) of pure protected taxoid 7-Triethylsilyl-10-acetyl-2'-triisopropylsilyl-3'-desphenyl-3'-(1-propenyl )docetaxel (la-P) was collected (90% conversion, 88% conversion yield) as a white solid: Mp. 131.0-132.5°C; 1 H NMR (CDCl3, 250 MHz) δ0.57 (q, J=7.7 Hz, 6H), 0.92 (t, J=7.7 Hz, 9H), 1.05-1.11 (m, 21H), 1.20 (s, 3H), 1.23 (s, 3H), 1.32 (s, 9H), 1.69 (s, 3H), 1.73 (d, J=6.2 Hz, 3H), 1.76-1.95 (m, 1H), 2.01 (s, 3H), 2.18 (s, 3H), 2.22-2.35 (m, 2H), 2.41 (s, 3H), 2.43-2.60 (m, 1H), 3.83 (d, J=6.8 Hz, 1 H), 4.17 (d, J=8.3 Hz, 1 H), 4.31 (d, J=8.3 Hz, 1 H), 4.42-4.55 (m, 2H), 4.62 (br m, 1 H), 4.85-4.98 (m, 2H), 5.46 (dd, J=14.3, 6.2 Hz, 1 H), 5.62-5.75 (m, 2H), 6.18 (t, J=9.1 Hz, 1H), 6.47 (s, 1H), 7.49 (t, J=7.2 Hz, 2H), 7.59 (t, J=7.2 Hz, 1H), 8.11 (d, J=7.2 Hz, 2H); 13 C NMR (63 MHz, CDCl3) δ5.29, 6.71, 10.04, 12.50, 14.41, 17.71, 17.94, 20.85, 21.24, 22.75, 26.39, 28.18, 35.36, 37.21, 43.28, 46.73, 55.0, 58.21, 71.23. 72.24, 74.89, 75.06, 78.05, 79.5, 81.12, 84.24, 127.67, 128.64, 129.25, 130.19, 133.40, 133.53, 140.74, 155.0, 167.0, 169.25, 169.89, 171.64, 203.72.

In a similar manner, the following 7-triethylsilyl-10-O-substituted 2'-triisopropylsilyl-3'-(1-propenyl)docetaxels (I-P) were obtained in high yields:

7-Triethylsilyl-10-propanoyl-2'-triisopropylsilyl-3'-desphenyl-3'-(2-methyl -1-propenyl)docetaxel (Ib-P):

1 H NMR (CDCl3, 250 MHz) δ0.53 (q, 6 H), 0.86 (t, 9 H), 1.09 (s, 21 H), 1.15 (s, 3H), 1.19-1.20 (m, 6 H), 1.31 (s, 9 H), 1.66 (s, 3 H), 1.73 (S, 3 H), 1.77 (s, 3 H), 1.86 (m, 1 H), 1.91 (s, 3 H), 2.34 (s, 3 H), 2.38 (s, 2 H), 2.41 (m, 1 H), 3.81 (d, J=6.6 Hz, 1 H), 4.15 (AB, JAB =8.1 Hz, 1 H), 4.26 (AB, JAB =8.1 Hz, 1 H), 4.41 (s, 1 H), 4.45 (m, 1 H), 4.79 (m, 1 H+NH), 4.89 (d, J=8.9 Hz, 1 H), 5.30 (d, J=7.7 Hz, 1 H), 5.65 (d, J=6.6 Hz, 1 H), 6.06 (t, 1 H), 6.47 (s, 1 H), 7.40 (t, 2 H), 7.54 (t, 1 H), 8.06 (d, 2 H).

7-Triethylsilyl-10-cyclopropanecarbonyl-2'-triisopropylsilyl-3'-desphenyl-3 '-(2-methyl-1-propenyl) docetaxel (Ic-P):

1 H NMR (CDCl3, 300 MHz) δ0.42 (m, 6 H), 0.82 (m, 9 H), 1.04 (s, 21 H), 1.12 (s, 3 H), 1.16 (s, 3 H), 1.18 (m, 2 H), 1.27 (s, 6 H), 1.62 (s, 3 H), 1.68 (bs, 5 H), 1.72 (s, 3 H), 1.84 (dt, 1 H), 1.94 (s, 3 H), 2.28 (s, 3 H), 2.32 (s, 2 H), 2.88 (ddd, 1 H), 2.50 (d, 1 H), 3.76 (d, J=6.9 Hz, 1 H), 4.11 (AB, JAB =8.4 Hz, 1 H), 4.22 (AB, JAB =8.4 Hz, 1 H), 4.36 (bs, 1 H), 4.39 (m, 1 H), 4.69 (m, 1 H+NH), 4.85 (d, J=9.0 Hz, 1 H), 5.25 (d, J=8.1 Hz, 1 H), 5.61 (d, J=6.6 Hz, 1 H), 5.99 (t, 1 H), 6.41 (s, 1 H), 7.37 (t, 2 H), 7.51 (t, 1 H), 8.02 (d, 2 H).

7-Triethylsilyl-10-crotonoyl-2'-triisopropylsilyl-3'-desphenyl-3'-(2-methyl -1-propenyl)docetaxel (Id-P):

1 H NMR (CDCl3, 250 MHz) δ0.51 (q, 6 H), 0.87 (t, 9 H), 1.10 (s, 21 H), 1.17 (s, 3 H), 1.24 (s, 3 H), 1.32 (s, 9 H), 1.68 (s, 3 H), 1.74 (s, 3 H), 1.78 (s, 3 H), 1.86 (m, 1 H), 1.90 (d, 3 H), 2.03 (s, 3 H), 2.35 (s, 3 H), 2.39 (s, 2 H), 2.45 (m, 1 H), 3.84 (d, J=7.1 Hz, 1 H), 4.17 (AB, JAB =8.3 Hz, 1 H), 4.28 (AB, JAB =8.3 Hz, 1 H), 4.42 (d, 1 H), 4.45 (dd, J=6.4 Hz, 10.2 Hz, 1 H), 4.75 (m, 1 H+NH), 4.91 (d, J=8.5 Hz, 1 H), 5.31 (d, J=8.2 Hz, 1 H), 5.67 (d, J=7.1 Hz, 1 H), 5.92 (d, 1 H), 6.04 (t, 1 H), 6.51 (s, 1 H), 6.99 (m, 1 H), 7.42 (t, 2 H), 7.56 (t, 1 H), 8.08 (d, 2 H).

7-Triethylsilyl-10-N,N-dimethylcarbamoyl-2'-triisopropylsilyl-3'-desphenyl- 3'-(2-methyl-1-propenyl)docetaxel (Ie-P):

1 H NMR (CDCl3, 250 MHz) δ0.52 (m, 6 H), 0.86 (m, 9 H), 1.09 (s, 21 H), 1.17 (s, 3 H), 1.19 (s, 3 H), 1.31 (s, 6 H), 1.66 (s, 3 H), 1.72 (s, 3 H), 1.76 (s, 3 H), 1.85 (dt, 1 H), 2.03 (s, 3 H), 2.33 (s, 3 H), 2.38 (s, 3 H), 2.48 (ddd, 1 H), 2.91 (s, 3 H), 3.03 (s, 3 H), 3.82 (d, J=6.9 Hz, 1 H), 4.15 (AB, JAB =8.2 Hz, 1 H), 4.25 (AB, JAB =8.2 Hz, 1 H), 4.39 (m, 1 H), 4.41 (bs, 1 H), 4.73 (m, 1 H+NH), 4.89 (d, J=8.8 Hz, 1 H). 5.30 (d, J=8.0 Hz, 1 H), 5.65 (d, J=6.9 Hz, 1 H), 6.04 (t, 1 H), 6.38 (s, 1 H), 7.39 (t, 2 H), 7.54 (t, 1 H), 8.06 (d, 2 H).

7-Triethylsilyl-10-methoxycarbonyl-2'-triisopropylsilyl-3'-desphenyl-3'-(2- methyl-1-propenyl) docetaxel (If-P):

1 H NMR (CDCl3, 250 MHz) δ0.52 (m, 6 H), 0.88 (m, 9 H), 1.10 (s, 21 H), 1.18 (s, 6 H), 1.32 (s, 9 H), 1.68 (s, 3 H), 1.73 (s, 3 H), 1.77 (s, 3 H), 1.83 (dt, 1 H), 2.00 (s, 3 H), 2.34 (s, 3 H), 2.38 (s, 2 H), 2.44 (ddd, 1 H), 3.79 (d, 1 H), 3.80 (s, 3 H), 4.15 (AB, JAB =8.3 Hz, 1 H), 4.27 (AB, JAB =8.3 Hz, 1 H), 4.41 (d, J=2.3 Hz, 1 H), 4.44 (m, 1 H), 4.74 (m, 1 H+NH), 4.90 (d, J=8.3 Hz, 1 H), 5.30 (d, J=8.2 Hz, 1 H), 5.64 (d, J=7.0 Hz, 1 H), 6.04 (t, 1 H), 6.26 (s, 1 H), 7.40 (t, 2 H), 7.55 (t, 1 H), 8.06 (d, 2 H).

7-Triethylsilyl-10-acryloyl-2'-triisopropylsilyl-3'-desphenyl-3'-(2-methyl- 1-propenyl)docetaxel (Ig-P):

1 H NMR (CDCl3, 250 MHz) δ0.51 (m, 6 H), 0.86 (m, 9 H), 1.10 (s, 21 H), 1.16 (s, 3 H), 1.24 (s, 3 H), 1.32 (s, 9 H), 1.62 (s 3 H), 1.68 (s, 3 H), 1.74 (s, 3 H), 1.78 (s, 3 H), 1.83 (m, 1 H), 2.35 (s, 2 H), 2.39 (s, 3 H), 2.42 (m, 1 H), 3.83 (d, J=7.3 Hz, 1 H), 4.16 (AB, JAB =8.3 Hz, 1 H), 4.28 (AB, JAB =8.3 Hz, 1 H), 4.41 (d, J=2.1 Hz, 1 H), 4.45 (m, 1 H), 4.74 (m, 1 H+NH), 4.90 (d, J=9.4 Hz, 1 H), 5.30 (d, J=7.8 Hz, 1 H), 5.62 (d, J=7.3 Hz, 1 H), 5.88 (d, J=10.3 Hz, 1 H), 6.04 (t, 1 H), 6.17 (m, 1 H), 6.46 (m, 1 H), 6.52 (s, 1 H), 7.41 (t, 2 H), 7.56 (t, 1 H), 8.07 (d, 2 H).

3'-Desphenyl-3'-(1-alkenyl)-10-O-substituted docetaxel (I):

To a solution of 46 mg. (0.042 mmol) of the protected taxoid Ia-P in 3 mL of 1:1 mixture of acetonitrile and pyridine was added 0.5 mL of HF/pyridine (70:30). The reaction mixture was stirred at 35-40°C for 2 hours. The reaction was quenched with 2N HCl. The mixture was extracted with EtOAc and the organic layer washed with 2N HCl and brine. After drying over MgSO4, the crude product was purified by flash chromatography on silica gel (hexane:EtOAc=1:2) to yield 24 mg (70% yield) of the pure taxoid 3'-desphenyl-3'-(1-propenyl)-10-acetyldocetaxel (Ia) as a white solid: Mp. 152.0-155.0°C; [α]D -86.7° (c, 0.15, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ1.15 (s, 3H), 1.25 (s, 3H), 1.32 (s, 9H), 1.67 (s, 3H), 1.75 (d, J=6.3 Hz, 3H), 1.86 (br s, 4H), 2.23 (s, 3H), 2.30-2.39 (m, 2H), 2.40 (s, 3H), 2.45-2.60 (m, 1H), 3.38 (br s, 1H), 3.81 (d, J=6.9 Hz, 1H), 4.17 (d, J=8.4 Hz, 1H), 4.30-4.33 (m, 2H), 4.42 (dd, J=10.5, 6.9 Hz, 1H), 4.60 (br m, 1H), 4.90-4.98 (m, 2H), 5.53 (dd, J=16.2, 6.3 Hz, 1H), 5.67 (d, J=6.9 Hz, 1H), 5.72-5.82 (m, 1H), 6.21 (t, J=8.8 Hz, 1H), 6.30 (s, 1H), 7.52 (t, J=7.2 Hz, 2H), 7.61 (t, J=7.2 Hz, 1H), 8.11 (d, J=7.1 Hz, 2H); 13 C NMR (63 MHz, CDCl3) δ9.53, 14.95, 17.87, 20.84, 21.82, 22.54, 26.69, 28.18, 35.45, 35.60, 54.90, 58.62, 72.19, 73.12, 74.98, 75.61, 79.03, 79.55, 81.10, 84.41, 127.37. 128.71, 129.1, 130.19, 133.1, 133.68, 142.50, 155.50, 167.20, 170.13, 171.5, 173.40, 203.73. Anal. Calcd. for C42 H55 O15 N: C, 61.98; H, 6.81; N, 1.72. Found: C, 62.12; H, 6.59; N, 1.67.

In a similar manner the following 3'-Desphenyl-3'-( 1-alkenyl)-10-O-substituted docetaxel (Ib-g) were obtained in high yields:

3'-Desphenyl-3'-(2-methyl-1-propenyl)-10-propanoyldocetaxel (Ib):

White solid; [α]D21 -40° (c 1.00, CHCl3); 1 H NMR (CDCl3, 300 MHz) δ1.08 (s, 3 H), 1.13-1.18 (m, 6 H), 1.28 (s, 9 H), 1.60 (s, 3 H), 1.69 (s, 6 H), 1.72 (m, 1 H), 1.83 (s, 3 H), 2.29 (s, 3 H), 2.31 (s, 2 H), 2.44 (m, 3 H), 3.38 (bs, OH), 3.74 (d, J=6.9 Hz, 1 H), 4.10 (AB, JAB =8.1 Hz, 1 H), 4.13 (bs, 1 H), 4.22 (AB, JAB =8.1 Hz, 1 H), 4.33 (dd, J=7.5 Hz, 10.1 Hz, 1 H), 4.67 (m, 1 H+NH), 4.88 (d, J=9.3 Hz, 1 H), 5.23 (d, J=8.4 HZ, 1 H), 5.59 (d, J=6.9 Hz, 1 H), 6.06 (t, 1 H), 6.24 (s, 1 H), 7.37 (t, 2 H), 7.51 (t, 2 H), 8.01 (d, 2 H); 13 C NMR (CDCl3, 63 MHz) δ9.0, 9.5, 14.9, 18.5, 21.8, 22.3, 25.7, 26.6, 27.5, 28.2, 35.5, 43.1, 45.6, 51.6, 55.5, 58.5, 72.1, 72.3, 73.7, 75.0, 75.4, 76.4, 76.5, 77.0, 77.5, 79.1, 79.9, 81.0, 84.3, 120.6, 128.6, 129.2, 130.1, 132.9, 133.6, 137.8, 142.4, 155.4, 166.9, 170.1, 173.0, 174.6, 203.8. HRMS (FAB, DCM/NBA), m/z: Calcd. for C44 H59 O15 NH+, 842.3962. Found, 842.4007.

3'-Desphenyl-3'-(2-methyl-1-propenyl)-10-cyclopropanecarbonyldocetaxel (Ic):

White solid; [α]D21 -160° (c 1.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ1.10 (m, 2 H), 1.14 (s, 3 H), 1.25 (s, 3 H), 1.34 (s, 9 H), 1.65 (s, 3 H), 1.71 (s, 2 H), 1.75 (s, 6 H), 1.84 (dt, 1 H), 1.88 (s, 3 H), 2.34 (s, 3 H), 2.37 (s, 2 H), 2.46 (ddd, 1 H), 2.56 (d, J=3.3 Hz, 1 H), 3.36 (d, OH), 3.78 (d, J=6.9 Hz, 1 H), 4.13 (d, J=8.4 Hz, 1 H), 4.18 (bs, 1 H), 4.27 (AB, JAB =8.4 Hz, 1 H), 4.40 (m, 1 H), 4.72 (m, 1 H+NH), 4.93 (AB, JAB =8.6 Hz, 1 H), 5.28 (d, J=7.6 Hz, 1 H), 5.64 (d, J=6.9 Hz, 1 H), 6.16 (t, 1 H), 6.28 (s, 1 H), 7.43 (t, 2 H), 7.56 (t, 1 H), 8.07 (d, 2 H); 13 C NMR (CDCl3, 63 MHz) δ9.1, 9.4, 9.5, 13.0, 14.9, 18.5, 21.9, 22.4, 25.7, 26.7, 28.2, 35.5, 35.6, 43.2, 45.6, 51.6, 58.5, 72.2, 72.3, 73.7. 75.0, 75.4. 76.5, 77.0, 77.5, 79.2, 79.7, 81.0, 84.4, 120.6, 128.6, 129.2, 130.1, 132.9, 133.6, 137.9, 142.6, 155.4, 166.9, 170.1, 175.1, 203.9; IR (neat, cm-1): 3368, 2989, 2915, 1786, 1754, 1725, 1709, 1641, 1630, 1355, 1315, 1109. HRMS (FAB, DCM/NBA/NaCl), m/z: Calcd. for C45 H59 O15 NNa+, 876.3784. Found 876.3782.

3'-Desphenyl-3'-(2-methyl-1-propenyl)-10-crotonoyldocetaxel (Id):

White solid; [α]D21 -30° (c 1.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ1.16 (s, 3 H), 1.26 (s, 3 H), 1.35 (s, 9 H), 1.67 (s, 3 H), 1.76 (s, 6 H), 1.22 (m, 1 H), 1.90 (s, 3 H), 1.92 (dd, 3 H), 2.35 (s, 3 H), 2.39 (s, 2 H), 2.49 (m, 1 H), 3.38 (bs, OH), 3.82 (d, J=6.9 Hz, 1 H), 4.10 (AB, JAB =8.3 Hz, 1 H), 4.20 (bs, 1 H), 4.29 (AB, JAB =8.3 Hz, 1 H), 4.45 (m, 1 H), 4.73 (m, 1 H+NH), 4.95 (d, J=7.9 Hz, 1 H), 5.30 (d, 1 H), 5.66 (d, J=6.9 Hz, 1 H), 5.95 (dd, 1 H), 6.14 (t, 1 H), 6.36 (s, 1 H), 7.03 (m, 1 H), 7.44 (t, 2 H), 7.57 (t, 1 H), 8.08 (d, 2 H); 13 C NMR (CDCl3, 63 MHz) δ9.5, 14.9, 18.2, 18.5, 21.9, 22.4, 25.7, 26.7, 28.2, 29.6, 35.5, 35.6, 43.2, 45.6, 51.6, 58.6, 72.2, 72.3, 73.7, 75.1, 75.3, 76.5, 77.0, 77.5, 79.2, 79.9, 81.0, 84.4, 120.6, 121.6, 128.6, 129.2, 130.1, 132.9. 133.6, 137.9, 142.6. 147.2, 155.4, 166.2, 166.9, 170.0, 173.0, 174.6, 203.8. HRMS (FAB, DCN/NBA/NaCl) m/z: Calcd. for C45 H59 O15 NNa+, 876.3782. Found, 876.3749.

3'-Desphenyl-3'-(2-methyl-1-propenyl)-10-N,N-dimethylcarbamoyldocetaxel (Ie):

White solid; [α]D21 -50° (c 2.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ1.13 (s, 3 H), 1.23 (s, 3 H), 1.33 (s, 9 H), 1.64 (s, 3 H), 1.74 (s, 6 H), 1.85 (dt, 1 H), 1.89 (s, 3 H), 2.33 (s, 3 H), 2.36 (s, 2 H), 2.45 (ddd, 1 H), 2.93 (s, 3 H), 3.02 (s, 3 H), 3.20 (bs, Oh), 3.45 (d, OH), 3.78 (d, J=6.9 Hz, 1 H), 4.14 (AB, JAB =8.4 Hz, 1 H), 4.18 (bs, 1 H), 4.26 (AB, JAB =8.4 Hz, 1 H), 4.40 (dd, J=6.7 Hz, 10.2 Hz, 1 H), 4.69 (d, 1 H), 4.80 (s, NH), 4.93 (d, J=8.6 Hz, 1 H), 5.27 (d, J=7.6 Hz, 1 H), 5.62 (d, J=6.9 Hz, 1 H), 6.12 (t, 1 H), 6.23 (s, 1 H), 7.41 (t, 2 H), 7.55 (t, 1 H), 8.06 (d, 2 H); 13 C NMR (CDCl3, 63 MHz) δ9.3, 15.0, 18.5, 22.2, 22.3, 25.7, 26.8, 28.2, 35.3, 35.6, 36.0, 36.6, 43.1, 45.6, 51.6, 58.4, 72.3, 72.4, 73.7, 75.2, 76.2, 76.4, 76.5, 77.0, 77.5, 79.2, 81.0, 84.6, 128.6, 129.2, 130.1, 133.1, 133.6, 137.8, 142.9, 155.4, 156.1, 166.9, 170.0, 173.0, 205.6. HRMS (FAB, DCM/NBA) m/z: Calcd. for C44 H60 O15 N2 Na+, 879.3891. Found, 879.3870.

3'-Desphenyl-3'-(2-methyl-1-propenyl)-10-methoxycarbonyldocetaxel (If):

White solid; [α]D21 -15.0° (c 2.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ1.14 (s, 3 H), 1.23 (s, 3 H), 1.33 (s, 9 H), 1.68 (s, 3 H), 1.71 (s, 6 H), 1.87 (m, 1 H), 1.92 (s, 3 H), 2.34 (s, 3 H), 2.47 (d, 2 H), 2.55 (m, 1 H), 3.40 (bs, OH), 3.76 (d, J=6.9 Hz, 1 H), 3.85 (s, 3 H), 4.15 (AB, JAB =8.3 Hz, 1 H), 4.19 (bs, 1 H), 4.28 (AB, JAB =8.3 Hz, 1 H), 4.38 (m, 1 H), 4.72 (m, 1 H+NH), 4.93 (d, J=8.6 Hz, 1 H), 5.29 (d, J=7.8 Hz, 1 H), 5.64 (d, J=6.9 Hz, 1 H), 6.11 (s, 1 H), 6.15 (s, 1 H), 7.43 (t, 2 H), 7.56 (t, 1 H), 8.07 (d, 2 H); 13 C NMR (CDCl3, 75 MHz) δ9.4, 15.0, 18.5, 21.7, 22.3, 25.7, 26.5, 28.2, 35.5, 43.1, 45.6. 51.6, 55.5. 58.6, 72.0, 72.2, 73.7, 75.0, 76.4, 76.5, 77.0, 77.2, 77.4, 78.3, 79.1, 79.9, 81.0, 84.3, 120.6, 128.6, 129.2, 130.1, 132.5, 133.6, 137.9, 143.4, 155.4, 155.7, 166.9, 170.1, 172.9. 203.9. HRMS (FAB, DCM/NBA/PPG) m/z: Calcd. for C43 H57 O16 NH+, 844.3710. Found, 844.3755.

3'-Desphenyl-3'-trifluoromethyl-10-acetyldocetaxel (Ig):

White solid; 1 H NMR (250 MHz CDCl3): δ1.14 (s, 3 H), 1.24 (S, 3 H), 1.30 (s, 9 H), 1.67 (s, 3 H), 1.75 (br s, 1 H), 1.92 (s, 3 H), 2.24 (s, 3 H), 2.28-2.37 (m, 5 H), 2.48-2.61 (m, 1 H), 3.46 (br d, 1 H), 3.79 (d, J=7.0 Hz, 1 H), 4.16 (d, J=8.3 Hz, 1 H), 4.30 (d, J=8.3 Hz, 1 H), 4.39 (br t, 1 H), 4.71-4.84 (m, 2 H), 4.93 (d, J=8.1Hz, 1 H), 5.24 (d, J=10.6 Hz, 1 H), 5.65 (d, J=7.0 Hz, 1 H), 6.21-6.28 (m, 2 H), 7.49 (t, J=7.4 Hz, 2 H), 7.61 (t, J=7.4 Hz, 1 H), 8.11 (d, J=7.4 Hz, 2 H), 13 C NMR (63 MHz, CDCl3) δ9.57, 14.82, 20.86, 21.92, 22.35, 26.72, 27.94, 35.39, 35.58, 43.25, 45.65, 53.54, 54.03, 58.59, 68.08, 73.15, 73.32, 74.94, 75.48, 76.51, 79.01, 81.18, 81.37, 84.43, 126.24, 128.77, 129.01, 130.23, 133.38, 133.74, 141.60, 154.67, 167.16, 170.28, 171.25, 171.76, 203.54. Anal. Calc. for C40 H50 F3 NO15 :C, 57.07; H, 5.99; N, 1.66. Found: C, 56:33; H, 6.02; N, 1.69.

3'-Desphenyl-3'-(2-methylpropyl)-10-O-substituted docetaxel (Ib'):

A solution of 14 mg (0.016 mmol) of Ib in 2.0 mL of ethyl acetate was stirred under one atmosphere of hydrogen at room temperature, in the presence of palladium (10%) on activated carbon (23 mg). After 24 hours the suspension was purified by chromatography on silica gel (EtOAc) to afford 14 mg (100%) of 3'-desphenyl-3'-(2-methylpropyl)-10-propanoyldocetaxel (Ib') as a white solid: [α]D21 -30° (c 1.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ0.96 (d, 6 H), 1.13 (s, 3 H), 1.22-1.27 (m, 6 H), 1.30 (s, 9 H), 1.63 (s, 3 H), 1.73 (s, 2 H), 1.82 (m, 1 H), 1.88 (s, 3 H), 2.36 (s, 3 H), 2.40 (s, 2 H), 2.46 (m, 1 H), 2.49 (m, 2 H), 3.25 (bs, OH), 3.79 (d, J=7.0 Hz, 1 H), 4.09 (AB, JAB =8.3 Hz, 1 H). 4.16 (bs, 1 H), 4.27 (AB, JAB =8.3 Hz, 1 H), 4.38 (dd, J=6.7 Hz, 10.2 Hz, 1 H), 4.57 (d, J=9.5 Hz, NH), 4.94 (d, J=8.0 Hz, 1 H), 5.64 (d, J=7.0 Hz, 1 H), 6.13 (t, 1 H), 6.30 (s, 1 H), 7.43 (t, 2 H), 7.56 (t, 1 H), 8.08 (d, 2 H); 13 C NMR (CDCl3, 63 MHz) δ9.0, 9.5, 14.9, 21.8, 21.9, 22.5, 23.2, 24.6, 26.5, 27.5, 28.1, 29.6, 35.5, 41.2, 43.1, 45.6, 51.3, 58.5, 72.1, 72.6, 73.0, 75.1, 75.4, 76.4, 76.5, 77.0, 77.5, 79.1, 79.7, 81.0, 84.4, 128.6, 129.2, 130.1, 132.9, 133.6. 142.4, 155.5, 166.9, 169.9, 173.9, 174.6, 203.8. HRMS (FAB, DCM/NBA) m/z: Calcd. for C44 H61 O15 NH+, 844.4119. Found, 844.4157.

In the same manner, the following 3'-desphenyl-3'-(2-methylpropyl)-10-O-substituted docetaxel (Ic'-f') were obtained in quantitative yields:

3'-Desphenyl-3'-(2-methylpropyl)-10-cyclopropanecarbonyldocetaxel (Ic'):

White solid; [α]D21 -30° (c 1.00, CHCl3, 250 MHz) δ0.96 (d, 6 H), 1.09 (m, 2 H), 1.14 (s, 3 H), 1.24 (s, 3 H), 1.30 (s, 9 H), 1.62-1.70 (m, 4 H), 1.66 (s, 3 H), 1.73 (m, 1 H), 1.88 (s, 3 H), 2.36 (s, 3 H), 2.39 (s, 1 H), 2.48 (ddd, 1 H), 2.50 (d, 1 H), 3.20 (d, OH), 3.78 (d, J=6.9 Hz, 1 H), 4.16 (AB, JAB =8.3 Hz, 1 H), 4.20 (bs, 1 H), 4.27 (AB, JAB =8.3 Hz, 1 H), 4.40 (m, 1 H), 4.55 (d, NH), 4.93 (d, J=8.1 Hz, 1 H), 5.64 (d, J=7.0 Hz, 1 H), 6.14 (t, 1 H), 6.29 (s, 1 H), 7.43 (t, 2 H), 7.56 (t, 1 H), 8.09 (d, 2 H);, 3 C (CDCl3, 63 MHz) δ9.1, 9.4, 9.5, 13.0, 14.9, 21.9, 22.0, 22.5, 23.2, 24.7, 26.6, 28.1, 35.4, 35.5, 41.2, 43.1, 45.6, 51.3, 58.5, 72.2, 72.7, 72.9, 75.1, 75.4, 76.5, 77∅ 77.5. 79.2, 79.7, 81.0, 84.4, 128.6, 129.2, 130.2, 132.9, 133.6, 142.6, 155.5, 166.9, 169.9, 173.9, 175.1, 203.9. HRMS (FAB, DCM/NBC/NaCl), m/z: Calcd. for C45 H61 O15 NNa+, 878.3938. Found, 878.3926.

3'-Desphenyl-3'-(2-methylpropyl)-10-N,N-dimethylcarbamoyldocetaxel (Ie'):

White solid; [α]D21 -80° (c 2.00, CHCl3), 1 H NMR (CDCl3, 250 MHz) δ0.95 (d, 6 H), 1.14 (s, 3 H), 1.23 (s, 3 H), 1.29 (s, 9 H), 1.66 (s, 3 H), 1.68 (m, 2 H), 1.82 (m, 1 H), 1.90 (s, 3 H), 2.36 (s, 3 H), 2.39 (s, 2 H), 2.50 (m, 1 H), 2.95 (s, 3 H), 3.03 (s, 3 H), 3.22 (d, OH), 3.78 (d, J=7.0 Hz, 1 H), 4.10 (AB, JAB =8.3 Hz, 1 H), 4.16 (bs, 1 H), 4.27 (AB, JAB =8.3 Hz, 1 H), 4.41 (dd, J=6.5 Hz, 10.2 Hz, 1 H), 4.56 (d, NH), 4.95 (d, J=8.1 Hz, 1 H), 5.63 (d, J=7.0 Hz, 1 H), 6.14 (t, 1 H), 6.24 (s, 1 H), 7.42 (t, 2 H), 7.56 (t, 1 H), 8.08 (d, 2 H); 13 C NMR (CDCl3, 75 MHz) δ9.8, 15.3, 22.3. 22.7, 22.9, 23.6, 25.1, 27.2, 28.5, 35.8, 36.0, 36.4, 37.0, 41.6, 43.6, 46∅ 51.7, 58.9, 72.8, 73.1, 75.7, 76.6, 76.8, 76.9, 77.1, 77.4, 77.6, 77.8, 79.6, 80.0, 81.5, 85.0, 128.7, 129.0, 129.7, 130.6, 133.6, 133.9, 143.3, 155.9, 156.5, 167.3, 170.3, 174.3, 206∅ HRMS (FAB) m/z: Calcd. for C44 H62 O15 N2 Na+, 881.4074. Found, 881.4047.

3'-Desphenyl-3'-(2-methylpropyl)-10-methoxycarbonyldocetaxel (If'):

White solid; [α]D21 -70° (c 1.00, CHCl3); 1 H NMR (CDCl3, 250 MHz) δ0.96 (d, 6 H), 1.14 (s, 3 H), 1.23 (s, 3 H), 1.30 (s, 9 H), 1.66 (s, 2 H), 1.69 (s, 3 H), 1.84 /(m, 1 H), 1.92 (s, 3 H), 2.37 (s, 3 H), 2.47 (s, 2 H), 2.55 (m, 1 H), 3.24 (d, OH), 3.77 (d, J=6.8 Hz, 1 H), 3.86 (s, 3 H), 4.16 (AB, JAB =8.2 Hz, 1 H), 4.17 (bs, 1 H), 4.28 (AB, JAB =8.2 Hz, 1 H), 4.40 (dd, 1 H), 4.56 (d, J=9.3 Hz, NH), 4.94 (d, J=8.0 Hz, 1 H), 5.65 (d, J=7.0 Hz, 1 H), 6.11 (s, 1 H), 6.18 (t, 1 H), 7.43 (t, 2 H), 7.56 (t, 1 H), 8.09 (d, 2 H); 13 C NMR (CDCl3, 75 MHz) δ9.5, 15.0, 21.8, 22.5, 23.2, 24.7, 26.5, 28.1, 35.5, 35.6, 41.2, 43.0, 45.5, 51.3, 55.5, 58.5, 72.0, 72.6, 73.0, 75.0, 76.5, 77.0, 77.5, 78.3, 79.1, 79.7. 81.0, 84.3, 128.6, 129.2, 130.2, 132.5, 133.6, 143.5, 155.5, 155.7, 166.9, 170.0, 173.9, 204∅ HRMS (FAB, DCM/NBA) m/z: Calcd. for C43 H59 O16 NH+, 846.3912. Found, 846.3942.

Taxoid Ia and Ig were evaluated for tumor growth inhibitory activities against human tumor cell line, A121 (ovarian carcinoma), A549 (non-small cell lung carcinoma), HT-29 (colon carcinoma), MCF7 (mammary carcinoma) or MCF7-R (mammary carcinoma cells 180-fold resistant to adriamycin), after 72 h drug exposure according to the literature method (see below). Results are shown in Table 1. Lower numbers indicate higher potency. Paclitaxel, docetaxel, and RAH-1 (see above) were also used for comparison. The data represent the mean values of at least three separate experiments. Lower numbers indicate greater activity.

TABLE 1
______________________________________
A121a
A549a
HT-29a
MCF7a
Taxoid (ovarian) (NSCLC) (colon) (breast) MCF7-R
a
______________________________________
Paclitaxel
6.1 3.6 3.2 1.7 300
Docetaxel 1.2 1.0 1.2 1.0 235
RAH-1 1.4 0.45 0.96 0.54 113
Ia 0.90 0.54 0.76 0.51 14
Ig 0.37 0.25 0.4 0.25 17
______________________________________
a The concentration of compound which inhibits 50% (IC50, nM) o
the growth of human tumor cell line.

Assessment of cell growth inhibition was determined according to the methods of Skehan et al., J. Nat. Cancer Inst. 1990, 82, 1107. Briefly, cells were plated between 400 and 1200 cells/well in 96 well plates and incubated at 37°C for 15-18 h prior to drug addition to allow attachment of cells. Compounds tested were solubilized in 100% DMSO and further diluted in RPMI-1640 containing 10 mM HEPES. Each cell line was treated with 10 concentrations of compounds (5 log range). After a 72 h incubation, 100 mL of ice-cold 50% TCA was added to each well and incubated for 1 h at 4°C Plates were then washed 5 times with tap water to remove TCA, low-molecular-weight metabolites and serum proteins. Sulforhodamine B (SRB) (0.4%, 50 mL) was added to each well. Following a 5 min incubation at room temperature, plates were rinsed 5 times with 0.1% acetic acid and air dried. Bound dye was solubilized with 10 mM Tris Base (pH 10.5) for 5 min on a gyratory shaker. Optical density was measured at 570 nm.

Data were fit with the Sigmoid-Emax concentration-effect model (see Holford, N. H. G.; Scheiner, L. B., "Understanding the dose-effect relationship: Clinical applications of pharmaco-kinetic-pharmacodynamic models.", Clin. Pharimiacokin. 1981, 6, 429-453) with non-linear regression, weighted by the reciprocal of the square of the predicted response. The fitting software was developed by the Roswell Park Cancer Institute with Microsoft FORTRAN, and uses the Marquardt algorithm (see Marquardt, D. W., "An algorithm for least squares estimation of nonlinear parameters", J. Soc. Ind. Appl. Math. 1963, 11, 431-441) as adopted by Nash (see Nash, J. C., "Compact numerical method for computers: Linear algebra and function minimization", John Wiley & Sons, New York, 1979) for the non-linear regression. The concentration of drug which resulted in 50% growth inhibition (IC50) was calculated.

Since the new taxoids of this invention are unique in that these taxoids possess extremely high activities against drug-resistant human breast cancer cells MCF7-R (two orders of magnitude better than paclitaxel and docetaxel), the activities of these taxoids other than Ia and Ig were evaluated against human breast cancer cells (MCF7) (sensitive) and resistant cells (MCF7-R) (resistant) in the same manner as described above. Results are summarized in TABLE 2.

TABLE 2
______________________________________
MCF7 MCF7-R
Taxoid R1 R5 IC50 (nM) IC50 (nM)
______________________________________
Ib 2-methyl-1-propenyl
COCH2 CH3
0.21 2.16
Ib' 2-methylpropyl COCH2 CH3 0.35 2.84
Ic 2-methyl-1-propenyl cyclopropyl- 0.20 2.11
carbonyl
Ic' 2-methylpropyl cyclopropyl- 0.51 4.33
carbonyl
Id 2-methyl-1-propenyl crotonoyl 0.26 3.35
Ie 2-methyl-1-propenyl CON(CH3)2 0.13 4.91
Ie' 2-methylpropyl CON(CH3)2 0.36 5.80
If 2-methyl-1-propenyl CO2 CH3 0.14 5.25
If' 2-methylpropyl CO2 CH3 0.48 6.35
______________________________________

Ojima, Iwao

Patent Priority Assignee Title
10550093, Jul 01 2015 The Research Foundation for The State University of New York Third generation taxoids and methods of using same
11780956, Mar 13 2018 Eastman Chemical Company Build materials for additive manufacturing applications
6323205, Jul 07 1996 Sloan-Kettering Institute for Cancer Research Combinations of 10-propargyl-10-deazaaminopterin and taxols and methods of using same in the treatment of tumors
6500858, Oct 28 1994 SYNPLICITY, INC Taxoid anti-tumor agents and pharmaceutical compositions thereof
6515151, Dec 23 1992 Bristol-Myers Squibb Company Method for the preparation of novel sidechain-bearing taxanes and intermediates thereof
6596737, Feb 02 2000 FSU RESEARCH FOUNDATION, INC C10 carbamoyloxy substituted taxanes
6660866, Feb 02 2000 FSU RESEARCH FOUNDATION, INC C10 carbonate substituted taxanes
6835746, Oct 28 1994 The Research Foundation of State University of New York Taxoid anti-tumor agents and pharmaceutical compositions thereof
6906040, Sep 22 2000 Bristol-Myers Squibb Company Method for reducing toxicity of combined chemotherapies
6906088, Feb 02 2000 FSU Research Foundation, Inc. Taxanes having a C10 carbamoyloxy substituent
6916942, Feb 03 2000 Bristol-Myers Squibb Company Process for the preparation of C-4 carbonate taxanes
6927211, Sep 22 2000 Bristol-Myers Squibb Company Method for reducing toxicity of combined chemotherapies
7056946, Feb 02 2000 FSU Research Foundation, Inc. C10 carbonate taxane compositions
7256213, Feb 02 2000 Florida State University Research Foundation, Inc. Taxanes having a C10 carbonate substituent
7981926, Aug 12 2005 Research Foundation of State University of New York Fluorotaxoids
8835433, Feb 02 2010 Allos Therapeutics, Inc. Optically pure diastereomers of 10-Propargyl-10-deazaaminopterin and methods of using same for the treatment of cancer
9187481, Feb 02 2010 Allos Therapeutics, Inc. (2S)-2-[[4-[(1R)-1-[(2,4-diaminopteridin-6-yl)methyl]but-3-ynyl]benzoyl]amin]pentanedioic acid for the treatment of inflammatory disorders
9901578, Aug 17 2007 ALLOS THERAPEUTICS, INC Combination of 10-propargyl-10-deazaaminopterin and erlotinib for the treatment of non-small cell lung cancer
Patent Priority Assignee Title
5229526, Sep 23 1991 Florida State University Metal alkoxides
5294637, Jul 01 1992 Bristol-Myers Squibb Company Fluoro taxols
5399726, Jan 29 1993 Florida State University Process for the preparation of baccatin III analogs bearing new C2 and C4 functional groups
5728850, Sep 23 1991 Florida State University Taxanes having a butenyl substituted side-chain and pharmaceutical compositions containing them
5739362, Sep 23 1991 Florida State University Taxanes having an alkoxy, alkenoxy or aryloxy substituted side-chain and pharmaceutical compositions containing them
///
Executed onAssignorAssigneeConveyanceFrameReelDoc
Mar 04 1996The Research Foundation of State University of New York(assignment on the face of the patent)
Dec 20 1999RHONE-POULENC RORER S A AVENTIS PHARMA S A CHANGE OF NAME SEE DOCUMENT FOR DETAILS 0118550908 pdf
Jan 20 2000OJIMA, IWAONEW YORK RESEARCH FOUNDATION OF STATE UNIVERSITY OF, THEASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0105920631 pdf
Date Maintenance Fee Events
Sep 14 2000ASPN: Payor Number Assigned.
Nov 18 2003M1551: Payment of Maintenance Fee, 4th Year, Large Entity.
Dec 12 2003ASPN: Payor Number Assigned.
Dec 12 2003RMPN: Payer Number De-assigned.
Jan 11 2008M1552: Payment of Maintenance Fee, 8th Year, Large Entity.
Sep 21 2011M1553: Payment of Maintenance Fee, 12th Year, Large Entity.
Dec 20 2011LTOS: Pat Holder Claims Small Entity Status.


Date Maintenance Schedule
Aug 08 20034 years fee payment window open
Feb 08 20046 months grace period start (w surcharge)
Aug 08 2004patent expiry (for year 4)
Aug 08 20062 years to revive unintentionally abandoned end. (for year 4)
Aug 08 20078 years fee payment window open
Feb 08 20086 months grace period start (w surcharge)
Aug 08 2008patent expiry (for year 8)
Aug 08 20102 years to revive unintentionally abandoned end. (for year 8)
Aug 08 201112 years fee payment window open
Feb 08 20126 months grace period start (w surcharge)
Aug 08 2012patent expiry (for year 12)
Aug 08 20142 years to revive unintentionally abandoned end. (for year 12)